Small Molecule
C32H32O13S
29767-20-2
Refractory Lymphoblastic Leukemia, Acute, Childhood
Teniposide is a semi-synthetic derivative of podophyllotoxin, a naturally occurring lignan. It is classified as an antineoplastic agent and functions as a specific inhibitor of DNA topoisomerase II.[1] In the United States, its primary and sole approved indication is for induction therapy in combination with other anticancer agents for refractory childhood acute lymphoblastic leukemia (ALL).[4] Its application is broader in Europe, where it is also approved for the treatment of Hodgkin's lymphoma, various primary brain tumors, neuroblastoma, and other pediatric malignancies.[7]
The compound is identified by several names and codes, including its chemical name, the code name VM-26, and the trade names Vumon® and Vehem®.[6] A comprehensive summary of its identifiers and chemical properties is provided in Table 1.
Table 1: Drug Identification and Chemical Properties of Teniposide
| Property | Value | Source(s) |
|---|---|---|
| DrugBank ID | DB00444 | 4 |
| Type | Small Molecule | 4 |
| CAS Number | 29767-20-2 | 6 |
| IUPAC Name | (5S,5aR,8aR,9R)-5-dioxin-6-yl]oxy]-9-(4-hydroxy-3,5-dimethoxyphenyl)-5a,6,8a,9-tetrahydro-5H-benzofuro[6,5-f]benzodioxol-8-one | 3 |
| Molecular Formula | $C_{32}H_{32}O_{13}S$ | 6 |
| Molecular Weight | 656.65 - 656.7 g/mol | 6 |
| InChIKey | NRUKOCRGYNPUPR-QBPJDGROSA-N | 6 |
| Canonical SMILES | COC1=CC(=CC(=C1O)OC)[C@H]2[C@@H]3C@HO[C@H]6O | |
| Synonyms/Codes | VM-26, Vumon, Vehem, NSC 122819, EPT, PTG |
Teniposide's development originates from efforts to modify podophyllotoxin, a cytotoxic lignan extracted from the rhizome of the American mandrake plant, Podophyllum peltatum, to create derivatives with an improved therapeutic index. It is a glycoside of podophyllotoxin, specifically identified as 4'-demethylepipodophyllotoxin 9-(4,6-O-(R)-2-thenylidene-beta-D-glucopyranoside). The semi-synthetic manufacturing process involves a condensation reaction between thiophene-2-aldehyde and 4'-demethylepipodophyllotoxin.
Physically, Teniposide is a white to off-white crystalline solid. It has a melting point in the range of 242–246 °C. As a lipophilic compound, it is practically insoluble in water, with an estimated solubility of only 5.9 mg/L at 25 °C, and is also insoluble in ether. Conversely, it is very soluble in organic solvents such as acetone and dimethylformamide. For long-term stability, the powdered form of the drug should be stored at -20°C.
The primary antineoplastic mechanism of Teniposide is the inhibition of DNA topoisomerase II alpha, a critical enzyme involved in managing DNA topology during replication, transcription, and repair. Unlike classic cytotoxic agents such as anthracyclines, Teniposide does not intercalate into the DNA helix or bind strongly to DNA. Instead, its action is more specific: it binds to the topoisomerase II enzyme after the enzyme has created a transient double-strand break in the DNA. This forms a stable ternary complex involving Teniposide, the enzyme, and the cleaved DNA.
By stabilizing this "cleavable complex," Teniposide prevents the topoisomerase II enzyme from completing its catalytic cycle, specifically inhibiting the re-ligation of the broken DNA strands. The persistence and accumulation of these stabilized complexes result in dose-dependent single- and double-stranded DNA breaks, as well as DNA-protein cross-links. The cellular DNA damage response mechanisms are overwhelmed by these lesions, ultimately triggering the apoptotic cell death pathway.
Teniposide's cytotoxicity is cell-cycle specific. It exerts its maximum effect during the late S (DNA synthesis) and early G2 (pre-mitotic) phases of the cell cycle, thereby arresting cells and preventing their entry into mitosis. This phase specificity is a crucial consideration in the design of clinical chemotherapy schedules to maximize its efficacy against proliferating cancer cells. In addition to its primary mechanism, some evidence suggests a secondary effect involving the inhibition of oxidative phosphorylation, indicated by observed mitochondrial abnormalities and reduced NADH-linked respiration in treated cells.
Teniposide is structurally and mechanistically similar to etoposide; both are epipodophyllotoxin derivatives developed to improve upon the toxicity profile of the parent compound, podophyllotoxin. The sole structural distinction is the substitution on the glucopyranoside ring: Teniposide possesses a thienyl group, whereas etoposide has a methyl group. This seemingly minor chemical modification leads to significant differences in their pharmacological profiles.
In vitro, Teniposide is a more potent cytotoxic agent than etoposide, demonstrating approximately 3- to 10-fold greater activity in producing DNA damage and killing cancer cells. A key factor contributing to this superior potency is its enhanced cellular uptake. Teniposide is more readily transported into cells, leading to higher intracellular drug concentrations and, consequently, a greater capacity for cytotoxicity, even though its direct inhibitory effect on the topoisomerase II enzyme is not substantially greater than that of etoposide.
However, this superior in vitro potency does not directly translate to broader clinical superiority, a phenomenon that can be described as a "potency-bioavailability paradox." The clinical utility of Teniposide is significantly modulated by its unfavorable pharmacokinetic properties in vivo. It exhibits extremely high plasma protein binding, with over 99% of the drug bound to proteins like albumin. This extensive binding drastically reduces the concentration of free, pharmacologically active drug available to diffuse into tumor tissues. This effectively counteracts its higher intrinsic potency at the cellular level. In contrast, etoposide is less extensively protein-bound, resulting in a higher fraction of unbound drug. Furthermore, the early development of an oral formulation for etoposide was perceived as a major clinical advantage, contributing to its more widespread investigation and adoption. This divergence illustrates a critical principle in drug development: favorable pharmacokinetics and bioavailability are as crucial as molecular potency for achieving clinical success.
Absorption: Teniposide is administered exclusively via the intravenous route, resulting in 100% bioavailability.
Distribution: As a highly lipophilic compound, Teniposide distributes into tissues, but its distribution is heavily influenced by its extensive plasma protein binding (>99%). The steady-state volume of distribution is relatively small, reported as 3 to 11 L/m² in children and 8 to 44 L/m² in adults. In patients with hypoalbuminemia, the reduced protein binding can lead to an increased volume of distribution and a higher fraction of free drug, potentially increasing the risk of toxicity. Penetration across the blood-brain barrier is generally limited, although higher cerebrospinal fluid (CSF) concentrations have been observed in patients with existing brain tumors.
Metabolism: The liver is the primary site of Teniposide metabolism. The metabolic pathways include conversion to its active catechol metabolite, a reaction mediated in part by the cytochrome P450 enzyme CYP3A4, though this process occurs more slowly for Teniposide than for etoposide. Teniposide is also a known substrate of the P-glycoprotein (P-gp) efflux transporter, which can influence its distribution and contribute to drug resistance.
Excretion: Following intravenous infusion, plasma concentrations of Teniposide decline in a biexponential manner. It has a longer terminal elimination half-life of approximately 5 hours compared to etoposide. Elimination occurs through both renal and fecal routes. Reflecting its more extensive metabolism, only 10% to 20% of a dose is excreted unchanged in the urine, a significantly lower proportion than the 30% to 70% observed with etoposide.
The approved clinical uses of Teniposide vary significantly by regulatory region.
Formulation: Teniposide is supplied as a sterile, nonpyrogenic solution for injection in 50 mg/5 mL (10 mg/mL) ampules. The formulation is a nonaqueous medium containing several excipients, notably Cremophor® EL (polyoxyl 35 castor oil), which is used to solubilize the lipophilic drug, along with N,N-dimethylacetamide, benzyl alcohol, and dehydrated alcohol.
Preparation and Dilution: Prior to administration, Teniposide must be diluted with either 5% Dextrose Injection, USP, or 0.9% Sodium Chloride Injection, USP. The final concentration for infusion should be 0.1 mg/mL, 0.2 mg/mL, 0.4 mg/mL, or 1.0 mg/mL. Diluted solutions at concentrations of 0.1, 0.2, and 0.4 mg/mL are stable at room temperature for up to 24 hours. However, to minimize the risk of precipitation, the 1.0 mg/mL solution should be administered within 4 hours of preparation. Refrigeration of diluted solutions is not recommended.
Administration: Teniposide must be administered as a slow intravenous infusion over a period of at least 30 to 60 minutes. Rapid intravenous injection is strictly contraindicated due to the risk of severe hypotension, an effect potentially mediated by the Cremophor® EL excipient.
Representative Dosing Regimens:
Clinical studies have established the efficacy of Teniposide, particularly in heavily pretreated and refractory pediatric populations.
A pivotal study conducted at St. Jude Children's Research Hospital (SJCRH) enrolled nine children with ALL who had failed initial induction therapy with a cytarabine-based regimen. Treatment with a combination of Teniposide and cytarabine induced complete remission in three of these patients. The durability of these responses was notable, with remission durations of 30 weeks, 59 weeks, and, in one case, 13 years, underscoring its potential for long-term disease control in a highly refractory setting.
Another study at SJCRH evaluated Teniposide in 16 children with ALL that was refractory to standard vincristine/prednisone-containing regimens. The addition of Teniposide to vincristine and prednisone demonstrated clinical activity in this challenging patient group.
In the context of non-Hodgkin's lymphoma, a randomized trial compared a standard regimen (ACVP: doxorubicin, cyclophosphamide, vincristine, prednisolone) with a Teniposide-containing regimen (ACTP) and a combination of both (ACTVP). The results showed that the Teniposide regimen (ACTP) produced complete remission rates (46%) and survival outcomes comparable to the vincristine regimen (ACVP, 44% CR). Importantly, ACTP was not associated with the neurotoxicity seen with vincristine. However, the study also found that combining Teniposide and vincristine (ACTVP) did not improve efficacy but significantly increased both neurotoxicity and myelosuppression, providing important guidance on its optimal use in combination therapy.
The use of Teniposide requires careful consideration and dose adjustments in specific patient populations.
The clinical use of Teniposide is associated with a significant and predictable toxicity profile, with myelosuppression being the most critical dose-limiting factor. A summary of key adverse reactions is provided in Table 2.
Table 2: Summary of Key Adverse Reactions to Teniposide
| System Organ Class | Adverse Reaction | Frequency / Notes | Source(s) |
|---|---|---|---|
| Hematologic | Myelosuppression (Dose-Limiting) | Very Common | |
| Neutropenia | 95%; Nadir at 7-14 days, recovery by 14-21 days | ||
| Thrombocytopenia | 80-85%; Nadir at 7-14 days, recovery by 14-21 days | ||
| Anemia | 88% | ||
| Leukopenia | 65-89% | ||
| Immune System | Hypersensitivity Reaction | 1-5% (Common); Can be life-threatening (anaphylaxis-like) | |
| Symptoms: chills, fever, tachycardia, bronchospasm, dyspnea, hypotension/hypertension, rash, flushing | |||
| Gastrointestinal | Mucositis / Stomatitis | 76% (Very Common) | |
| Nausea and Vomiting | 10-30% (Common) | ||
| Diarrhea | Common | ||
| Anorexia | Common | ||
| Skin and Subcutaneous Tissue | Alopecia | 9% (Common); Reversible | |
| Rash | Common | ||
| Cardiovascular | Hypotension | 2% (Uncommon); Associated with rapid infusion | |
| General Disorders and Administration Site | Extravasation Hazard | Irritant; can cause tissue necrosis | |
| Phlebitis | Possible at IV site | ||
| Nervous System | Peripheral Neurotoxicity | Reported | |
| Acute CNS Depression | Reported, especially with high doses | ||
| Neoplasms (Long-term) | Secondary Leukemia (t-ANLL) | 5-12%; Risk is schedule- and dose-dependent |
The FDA label for Teniposide carries a black box warning highlighting its significant risks. The warning stipulates that:
Contraindications for the use of Teniposide include:
The most serious long-term consequence of Teniposide therapy is the risk of developing a secondary malignancy, most commonly therapy-related acute nonlymphocytic leukemia (ANLL) or myelodysplastic syndrome (MDS). This risk exemplifies the "double-edged sword" nature of genotoxic chemotherapy. The very mechanism that confers its therapeutic efficacy—the induction of DNA double-strand breaks in cancer cells—is also responsible for its carcinogenic potential. Sublethal DNA damage inflicted upon healthy hematopoietic stem and progenitor cells can lead to mutations and chromosomal aberrations. Over time, these genetic insults can drive clonal expansion and malignant transformation.
The risk is not uniform and is highly dependent on the treatment schedule and cumulative dose. Studies have shown that prolonged, low-dose maintenance schedules (e.g., weekly or twice-weekly administration) are associated with a significantly higher risk of secondary leukemia, with a relative risk approximately 12 times greater than that observed in patients treated with other regimens. In contrast, short, intensive courses used for remission induction and consolidation have not been associated with the same elevated risk. This observation underscores that the frequency and chronicity of DNA insults to the stem cell pool are critical determinants of leukemogenesis.
Teniposide's carcinogenic potential is supported by its genotoxicity profile. It is mutagenic in the Ames test and is a potent clastogen, inducing DNA damage and chromosomal aberrations in a variety of in vitro and in vivo mammalian cell assays.
Pregnancy: Teniposide is classified as FDA Pregnancy Category D, indicating positive evidence of human fetal risk. The drug should not be used during pregnancy unless the potential benefit to the mother justifies the potential risk to the fetus. Animal studies have demonstrated that Teniposide is both teratogenic and embryotoxic, causing major fetal anomalies including spinal and rib defects and deformed extremities. Women of childbearing potential must be counseled on the risks and advised to use effective contraception throughout the treatment period.
Lactation: It is unknown whether Teniposide or its metabolites are excreted in human milk. Given the potential for serious adverse reactions in a nursing infant, breastfeeding is not recommended during therapy.
Male Fertility: Preclinical studies indicate that Teniposide can impair male fertility by causing a decrease in sperm count, genetic damage to sperm, and reduced testicular weight. Male patients of reproductive age should be informed of the risk of infertility and counseled on the option of sperm banking prior to initiating therapy.
The clinical use of Teniposide requires careful management of potential drug-drug interactions, which can be categorized as either pharmacokinetic or pharmacodynamic.
Interactions primarily involve the modulation of CYP3A4 metabolism and P-glycoprotein (P-gp) transport.
These interactions involve additive or synergistic effects on physiological systems.
Table 3: Clinically Significant Drug-Drug Interactions with Teniposide
| Interacting Agent/Class | Mechanism | Clinical Consequence | Management Recommendation |
|---|---|---|---|
| Strong CYP3A4 Inhibitors (e.g., ketoconazole, clarithromycin) | Pharmacokinetic (Inhibition of metabolism) | Increased Teniposide concentration and toxicity | Avoid or Use Alternate Drug; Monitor closely for toxicity if co-administration is unavoidable |
| Strong CYP3A4 Inducers (e.g., carbamazepine, apalutamide) | Pharmacokinetic (Induction of metabolism) | Decreased Teniposide concentration and efficacy | Avoid or Use Alternate Drug; Monitor for lack of efficacy |
| P-gp Inhibitors (e.g., cyclosporine, amiodarone) | Pharmacokinetic (Inhibition of efflux) | Increased Teniposide exposure and toxicity | Use Caution/Monitor |
| Anticoagulants / Antiplatelets (e.g., warfarin, abciximab, NSAIDs) | Pharmacodynamic (Additive effect) | Increased risk of severe bleeding | Monitor platelet counts and signs of bleeding; Avoid NSAIDs |
| Other Myelosuppressive Agents | Pharmacodynamic (Additive effect) | Increased severity and duration of bone marrow suppression | Monitor blood counts frequently; Dose adjustments may be necessary |
| Live Virus Vaccines (e.g., MMR, Varicella, Yellow Fever) | Pharmacodynamic (Impaired immune response) | Risk of disseminated, life-threatening infection | Contraindicated during and for at least 3 months after therapy |
The story of Teniposide begins not in a modern laboratory, but with the historical use of podophyllin, a resin derived from the Podophyllum plant, in folk medicine for centuries. The highly potent but toxic active ingredient, podophyllotoxin, was isolated in the 19th century. Recognizing its antimitotic properties, researchers in the mid-20th century sought to create derivatives with a better therapeutic window. This work, conducted at Sandoz in the 1960s, led to the synthesis of two key epipodophyllotoxins: Teniposide (VM-26) and its analogue, etoposide (VP-16).
Based on impressive preclinical activity against murine leukemias, Teniposide was advanced into clinical trials in 1971. While early results were encouraging, particularly in refractory leukemias, the focus of clinical development soon shifted almost exclusively to etoposide. This pivot was driven in part by perceived advantages in etoposide's formulation, including the availability of an oral dosage form, which simplified administration. Consequently, for many years, Teniposide was considered an under-investigated agent, prompting calls in the late 1980s and early 1990s for its re-evaluation in lymphomas, leukemias, and small cell lung cancer, where it had shown initial promise.
The regulatory and market status of Teniposide is fragmented globally, reflecting its evolution as a legacy chemotherapeutic agent in an era of rapidly advancing oncology care.
This pattern of divergent regulatory fates illustrates the typical lifecycle of an older cytotoxic drug. Teniposide was developed when its significant toxicities were deemed acceptable for treating life-threatening pediatric cancers for which few alternatives existed. As the therapeutic landscape evolved with the introduction of newer, more targeted, and potentially safer agents (e.g., immunotherapies like blinatumomab for ALL), the clinical niche for Teniposide narrowed in markets like the U.S. and Australia. This likely rendered its continued marketing commercially unviable, leading to its withdrawal. In contrast, its broader indication profile or different healthcare economic models in parts of Europe may have sustained its availability for specific, highly refractory conditions where it still holds clinical value.
Despite its discontinuation in some major markets, Teniposide remains a subject of clinical investigation, suggesting its unique mechanism of action is still considered relevant. Current research is focused on repositioning this older drug within modern therapeutic paradigms, particularly in biomarker-selected populations and in novel combination regimens.
The future clinical role of Teniposide is unlikely to be as a broad-spectrum agent. Instead, these ongoing studies suggest a potential for its revitalization as a niche therapeutic. Its future may lie in its application for specific, molecularly defined cancer subtypes or as a component in innovative combinations that pair its potent cytotoxic effects with the mechanisms of modern immunotherapy and targeted drugs.
Teniposide is a potent, cell-cycle specific topoisomerase II inhibitor with established efficacy in refractory pediatric hematologic malignancies. Its development from a natural product, podophyllotoxin, into a semi-synthetic chemotherapeutic agent highlights a classic paradigm in drug discovery. Its pharmacological profile is defined by a paradox: while demonstrating superior in vitro potency and cellular uptake compared to its analogue etoposide, its clinical utility is tempered by unfavorable pharmacokinetics, particularly its extensive plasma protein binding.
The clinical application of Teniposide is characterized by a significant and manageable toxicity profile, dominated by severe myelosuppression and a risk of hypersensitivity reactions. The most concerning long-term risk is the development of therapy-related secondary leukemias, a direct consequence of its DNA-damaging mechanism of action. This risk underscores the critical need for careful patient selection and risk-benefit assessment, especially in pediatric populations with long life expectancies.
The drug's divergent global regulatory status—approved for broad use in Europe but discontinued in the United States and Australia—reflects the dynamic nature of the oncology market, where legacy cytotoxic agents are often supplanted by newer, more targeted therapies. Nonetheless, ongoing clinical trials exploring Teniposide in biomarker-selected populations and in combination with immunotherapy suggest a potential for its repositioning. The future of Teniposide may not be in widespread use but as a valuable tool for specific, hard-to-treat cancers where its potent and unique mechanism of action can still address a critical unmet medical need.
Published at: October 17, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.